You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Aniracetam: An Evidence-Based Model for Preventing the Accumulation of Amyloid-β Plaques in Alzheimer’s Disease

Abstract

 Alzheimer’s disease is the leading cause of dementia in the world. It affects 6 million people in the United States and 50 million people worldwide. Alzheimer’s disease is characterized by the accumulation of amyloid-β plaques (Aβ), an increase in tau protein neurofibrillary tangles, and a loss of synapses. Since the 1990s, removing and reducing Aβ has been the focus of Alzheimer’s treatment and prevention research. The accumulation of Aβ can lead to oxidative stress, inflammation, neurotoxicity, and eventually apoptosis. These insults impair signaling systems in the brain, potentially leading to memory loss and cognitive decline. Aniracetam is a safe, effective, cognitive-enhancing drug that improves memory in both human and animal studies. Aniracetam may prevent the production and accumulation of Aβ by increasing α-secretase activity through two distinct pathways: 1) increasing brain derived neurotrophic factor expression and 2) positively modulating metabotropic glutamate receptors. This is the first paper to propose an evidence-based model for aniracetam reducing the accumulation and production of Aβ.

Aniracetam is a known cognitive enhancer whose pharmacological mechanisms are not fully understood. The main metabolites of aniracetam are N-anisoyl-γ-aminobutyric acid (N-anisoyl-GABA), 2-pyrrolidinone and anisic acid [1]. Aniracetam modulates metabotropic glutamate receptors (mGluRs) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-sensitive glutamate receptors, and it increases cholinergic activity in the hippocampus, prefrontal cortex, and striatum [2]. It also protects against glutamate excitotoxicity [3]. Aniracetam, in combination with AMPA, increases brain-derived neurotrophic factor (BDNF) [4], an important trophic factor in the brain that supports healthy memory, neurogenesis, and synaptogenesis [5]. A recent study from 2023 found that aniracetam, when combined with perampanel, reduces inflammation, and increases BDNF [6]. Research in humans and animals finds aniracetam has excellent safety, tolerability, and few drug interactions, making it an ideal candidate for the prevention and treatment of Alzheimer’s disease (AD) [1, 7].

ALPHA-SECRETASE

The majority of AD drugs have attempted to treat and prevent AD by reducing Aβ [8]. Many of these drugs have attempted to inhibit β-secretase as a means of reducing Aβ production [9]. Unfortunately, clinical trials using this approach have failed. Also, because β-secretase and γ-secretases both have multiple substrates other than amyloid-β protein precursor (AβPP), they are not ideal for AD treatment.

Targeting α-secretase activity is attractive therapeutically for AD because increasing α-secretase cleavage of AβPP has multiple positive effects regarding AD pathology [10]. First, cleaving AβPP with α-secretase prevents Aβ liberation. Second, α-secretase cleavage of AβPP produces sAβPPα, which is known to be neuroprotective [11]. Specifically, sAβPPα plays important roles in neuronal plasticity and survival: 1) it protects hippocampal neurons against excitotoxicity, 2) it protects neurons from Aβ toxicity, and 3) it protects neurons from hypoglycemic damage [12]. Third, there may be other neuroprotective downstream effects of upregulating α-secretase activity. For example, α-secretase is involved in the regulation of pro-inflammatory cytokines [13], and increasing α-secretase may reduce inflammation. Inflammation is a well-established risk factor for AD [14]. Because of the multiple neuroprotective effects of α-secretase, enhancing α-secretase activation is likely to help prevent AD [15].

It is important to note that α-secretase has multiple downstream substrates, and little is known about the signaling pathways that may stimulate α-secretase cleavage of AβPP [16].

Evidence suggests that aniracetam has the potential to increase α-secretase activity, thereby reducing Aβ production, via two distinct pathways: first, by increasing activity of BDNF, and second, by positively modulating metabotropic glutamate receptors (mGluRs). Both BDNF and positive modulation of mGluRs increase α-secretase activity and decrease Aβ [17–19].

AMYLOID-β PLAQUES

Excess Aβ is believed to be a significant contributor to the dysfunction that occurs in AD [20, 21]. Accumulation of Aβ damages neurons and synapses and often contributes to neuroinflammation [22, 23]. Specifically, Aβ can lead to oxidative stress, inflammation, neurotoxicity, and eventually apoptosis. These insults impair signaling systems in the brain, potentially leading to memory loss and cognitive decline [24, 25]. Aniracetam, a known nootropic and cognitive enhancer [1, 2, 26], has the potential to reduce Aβ by facilitating the non-amyloidogenic processing of AβPP by elevating α-secretase activity via increasing BDNF and modulating mGluRs.

Aβ is a 39 to 43 amino acid peptide derived from AβPP [27]. There are three known proteases that cleave AβPP: α-, β-, and γ-secretases. Aβ is created when AβPP is cleaved by β-secretase and γ-secretase [23]. When AβPP is cleaved at the beta, gamma, and caspase sites, the result is four peptides: sAβPPβ (soluble AβPP cleaved at the beta site), Aβ, Jcasp (the juxtamembrane peptide cleaved at the caspase site) and C31 (the final 31 amino acids of the protein) [28]. When AβPP is cleaved at the alpha site by α-secretase, the result is sAβPPα (soluble AβPP cleaved at the α site) and αCTF (carboxyterminal fragment), an 83-amino acid chain which is subsequently cleaved by γ-secretase producing AβPP intracellular domain (AICD) and P3 peptides [23, 29]. sAβPPα has neuroprotective properties [11, 30]. When AβPP is cleaved by α-secretase, the production of Aβ is prevented. Therefore, increasing α-secretase activity is a potential pathway for decreasing Aβ production and accumulation, as well as increasing neuroprotective sAβPPα. Indeed, brains of AD patients are deficient in α-secretase [31], a deficiency in α-secretase levels accelerates AD pathology [29], and increasing α-secretase activity decreases Aβ production [32, 33].

It is important to note that simply reducing Aβ is not necessarily beneficial to cognition or helpful in the treatment of AD. Many drugs that reduce Aβ have failed to benefit humans in clinical trials. Since 2018, nine drugs that directly reduced Aβ have failed in Phase III trials [8]. And not all drugs that elevate α-secretase activity lower Aβ in vivo or show benefit in clinical trials (e.g., Etazolate) [34]. There remain many unknowns in the field of AD research and Aβ biochemistry.

BRAIN DERIVED NEUROTROPHIC FACTOR

Aniracetam increases BDNF [4]. Aniracetam is a well-established positive modulator of AMPA receptors [35–39], specifically in the dentate gyrus and CA1 regions of the hippocampus [40]. Aniracetam also slows the desensitization of AMPA receptors and enhances synaptic plasticity [36, 41]. Aniracetam administered with AMPA increases BDNF release and enhances BDNF gene expression; AMPA+aniracetam increased BDNF levels 1.5-fold, and levels remained elevated 6 hours later [4]. Researchers have suggested that part of the efficacy of aniracetam is its ability to increase BDNF, likely through mGluR modulation and increased cholinergic activity [2].

There is a strong and consistent association between increased BDNF and reduced levels of Aβ [17, 18]. A 2017 study by Mark Mattson and colleagues found that BDNF reduces Aβ by enhancing a pathway that involves α-secretase. Specifically, researchers treated in vitro cultured cells with BDNF and found a significant reduction in both Aβ40 and Aβ42, compared to control cultures. Researchers concluded that BDNF reduces Aβ levels by increasing α-secretase activity. Interestingly, and surprisingly, their research found that increasing α-secretase cleavage of APP by increasing BDNF did not directly increase ADAM10 [16].

A 2022 study found that BDNF helps regulate AβPP processing, likely through α-secretase [42]. Indeed, treating cultures with BDNF decreased Aβ40 by 1.32-fold, decreased Aβ42 levels by 2.15-fold, and increased sAβPPα by 1.50-fold [42]. Retinoic acid, a trophic metabolite of vitamin A, when combined with BDNF, increases sAβPPα production. Investigators concluded that this increase was due to retinoic acid and BDNF shifting AβPP processing in favor of the neuroprotective α-secretase pathway [43].

Conversely, low levels of BDNF are associated with an increased risk of AD. Indeed, those with AD have significantly lower levels of BDNF in their blood than healthy controls [44]. Some researchers speculate that BDNF may also decrease BACE1, which would decrease β-secretase cleavage and reduce Aβ liberation [43].

BDNF has other neuroprotective properties relevant to AD outside of its ability to elevate α-secretase activity and reduce Aβ. BDNF plays a crucial role in supporting the function and survival of neurons that deteriorate in the advanced stages of AD. BDNF protects against excitotoxicity, promotes regeneration of dendrites, and reduces apoptosis [45, 46]. A 2023 study found that BDNF helps protect both mitochondria and neurons. Specifically, BDNF was found to improve mitochondrial function, protect neurons from oxidative stress, and protect dendrites [47]. This is highly relevant to AD because oxidative stress and degeneration of mitochondrial function is associated with cognitive decline and increased risk of AD [48–50].

In the human brain, BDNF exists in two forms: the BDNF precursor, proBDNF, and mature BDNF. Existing research has demonstrated a significant reduction in both proBDNF and mature BDNF in the brain during the late stages of AD. Specifically, proBDNF is reduced by 30% in AD brains, and mature BDNF is reduced by 62% in AD brains. This decrease in BDNF is also associated with a decline in cognition [51].

By enhancing BDNF expression, aniracetam shows promise as a potential treatment for AD by reducing Aβ plaque, enhancing mitochondrial function, and supporting the survival of neurons.

METABOTROPIC GLUTAMATE RECEPTORS

L-glutamate is the primary excitatory neurotransmitter in the central nervous system (CNS). mGluRs, which are classified as neuromodulatory receptors, offer a means for glutamate to regulate cell excitability and synaptic transmission through second messenger signaling pathways. Essentially, mGluRs modulate synaptic transmission and neuronal excitability throughout the CNS.

Glutamate and mGluRs may play an important role in AD [52]. Modulating mGluRs may help with AD in the following ways: 1) increase α-secretase activity and reduce Aβ formation, 2) protect against excitotoxicity, 3) reduce oxidative stress, and 4) enhance neuroplasticity [53, 54].

Aniracetam has the potential to reduce Aβ plaques by enhancing α-secretase activity by increasing mGluR activity. Aniracetam potentiates mGluR activity [2, 3, 55], and mGluR activation increases AβPP processing into non-amyloidogenic AβPPs in the hippocampus in rats [19]. Though other evidence suggests mGluRs stimulation may increase Aβ plaques [56].

The neurochemistry of aniracetam is not well understood, and it is not known which specific mGluRs aniracetam modulates. Group I mGluRs activation increases α-secretase activity [57]. Group III mGluRs activation facilitates non-amyloidogenic cleavage of AβPP, may increase BDNF levels, and helps remove extracellular Aβ via glial phagocytosis [58]. Activating group III mGluRs may also increase α-secretase and inhibit β-secretase expression [59]. Interestingly, downregulating Group 5 mGluRs may have neuroprotective effects and may be helpful in the treatment of AD [60].

Indeed, a 2020 article in the Journal of Alzheimers Disease detailed the therapeutic potential of modulating mGluRs for the treatment and prevention of AD [53].

SIDE EFFECTS

Aniracetam is well-tolerated in most clinical trials and does not increase liver enzymes [1]. However, it is not without side effects. The most common adverse events reported with aniracetam use were unrest, anxiety, uneasiness, and insomnia. Other unwanted side effects include urinary urgency, headache, vertigo, mild stomach pain, nausea, diarrhea, and rash. In clinical trials these effects were considered mild and did not necessitate withdrawal from the study [1].

Aniracetam is well-tolerated in the majority of clinical trials. One study of 109 elderly subjects took aniracetam or placebo for six months. Researchers wrote, “Tolerability of aniracetam was excellent” [61]. Another study of 115 subjects took aniracetam or placebo for six months. Researchers reported excellent tolerability [62].

Despite the high tolerability in clinical trials, healthcare practitioners are reporting unpleasant side effects in patients taking aniracetam. If the side effects of aniracetam are significantly impairing the quality of life, especially if they are impairing sleep—which is essential to brain health and memory consolidation—it is important for the patient and practitioner to determine if the cognitive-enhancing and neuroprotective benefits of aniracetam are worth enduring the side effects.

DISCUSSION

Aniracetam is a known cognitive enhancer and positive modulator of AMPA receptors and mGluRs. Multiple studies indicate that aniracetam likely increases α-secretase activity by increasing BDNF expression and positively modulating mGluRs. No research could be found in human or animal studies investigating the impact of aniracetam on Aβ accumulation or production. To the author’s knowledge, this paper is the first evidence-based model proposing that aniracetam lowers Aβ production and accumulation.

LIMITATIONS

There is much we do not know regarding AD neurobiochemistry. Researchers generally agree that cleaving AβPP with α-secretase will be neuroprotective by reducing Aβ. However, there remain multiple unknown aspects of AβPP, α-secretase, and Aβ biology. For example, which α-secretases effectively cleave AβPP to produce neuroprotective sAβPPα, and which ones do not? What are all of the downstream substrates of α-secretase, and what are the impacts of chronically elevating α-secretase? To what extent does modulating α-secretase affect levels of inflammation (e.g., TNF)? Clinical trials lasting six months report that aniracetam is well tolerated [61, 62], though this does not guarantee that there are no long-term negative impacts of chronically upregulating α-secretase.

Indeed, increasing α-secretase activity may be a double-edged sword. Cleaving AβPP with α- and γ-secretases does prevent the production of Aβ, and it also creates the peptides sAβPPα and P3. While sAβPPα has neuroprotective effects, P3 may be neurotoxic. P3 has demonstrated neurotoxic effects in vitro, specifically increasing neuronal apoptosis [63]. Recent research from 2020 found P3 to have amyloidogenic properties [64].

The biochemistry of aniracetam is not fully understood. Aniracetam modulates mGluRs, but does it modulate group I and/or group III mGluRs to the point of increasing α-secretase expression and increasing Aβ clearance? We do not know which α-secretase (e.g., ADAM9, ADAM10, ADAM19) aniracetam upregulates, nor which α-secretases cleave AβPP. Does aniracetam as a monotherapy, or used in tandem with other cholinergics or BDNF agonists, significantly reduce Aβ and slow down the progression of AD?

FUTURE RESEARCH

Because aniracetam has known cognitive and behavioral benefits in humans with mild cognitive impairment and early-stage AD [1, 61,65, 66], and aniracetam has a high level of tolerability and safety, human clinical trials could start immediately. I recommend starting with a 6-month crossover trial in adults with mild cognitive impairment. Ideally, researchers would measure cognition, serum BDNF levels, and Aβ using PET scans, at baseline and at every three months of the study. Subjects would be given 1,500 mg/day of aniracetam, taken with a source of fat to increase bioavailability (e.g., coconut oil, olive oil, or with food). To reduce potential side-effects, subjects could take a methylated B-complex vitamin and 1,200 mg of Alpha GPC with their aniracetam dose.

Further research is required to specify which mGluRs aniracetam modulates and which specific α-secretase or secretases aniracetam activates. Future animal studies could also investigate the downstream impact of chronically increasing α-secretase activity. I recommend starting by investigating if a daily dose of aniracetam (50 mg/kg) in rats significantly increases BDNF expression and which specific mGluRs groups aniracetam modulates.

AUTHOR CONTRIBUTIONS

Robert William Love (Conceptualization; Writing – original draft; Writing – review & editing).

ACKNOWLEDGMENTS

I want to thank my parents, Bob and Angie, for their years of love and support.

I would like to acknowledge the work of Dr. Dale Bredesen, Dr. Heather Sandison, and all researchers and neuroscientists investigating how to prevent Alzheimer’s disease. I want to thank Dr. Joshua Helman for providing feedback on this paper.

FUNDING

Funding was provided by Brain Fit For Life, LLC, a private company committed to helping improve brain health and reduce the prevalence of Alzheimer’s disease. Additional funding came from private donors. As of March 2024, Brain Fit For Life, LLC does not sell aniracetam.

CONFLICT OF INTEREST

The author has no conflict of interest to report.

DATA AVAILABILITY

All data are available in the main text.

REFERENCES

[1] 

Lee CR , Benfield P ((1994) ) Aniracetam: An overview of its pharmacodynamic and pharmacokinetic properties, and a review of its therapeutic potential in senile cognitive disorders. Drugs Aging 4: , 257–273.

[2] 

Shirane M , Nakamura K ((2000) ) Group II metabotropic glutamate receptors are a common target of N-anisoyl-GABA and 1S, 3R-ACPD in enhancing ACh release in the prefrontal cortex of freely moving SHRSP. Neuropharmacology 39: , 866–872.

[3] 

Pizzi M , Fallacara C , Arrighi V , Memo M , Spano P ((1993) ) Attenuation of excitatory amino acid toxicity by metabotropic glutamate receptor agonists and aniracetam in primary cultures of cerebellar granule cells. J Neurochem 61: , 683–689.

[4] 

Wu X , Zhu D , Jiang X , Okagaki P , Mearow K , Zhu G , McCall S , Banaudha K , Lipsky R , Marini A ((2004) ) AMPA protects cultured neurons against glutamate excitotoxicity through a phosphatidylinositol 3-kinase-dependent activation in extracellular signal-regulated kinase to upregulate BDNF gene expression. J Neurochem 90: , 807–818.

[5] 

Miranda M , Morici JF , Zanoni MB , Bekinschtein P ((2019) ) Brain-derived neurotrophic factor: A key molecule for memory in the healthy and the pathological brain. Front Cell Neurosci 13: , 363.

[6] 

Sharma H , Reeta KH , Sharma U , Suri V , Singh S ((2023) ) AMPA receptor modulation through sequential treatment with perampanel and aniracetam mitigates post-stroke damage in experimental model of ischemic stroke. Naunyn Schmiedebergs Arch Pharmacol 396: , 3529–3545.

[7] 

Endo H , Tajima T , Yamada H , Igata A , Yamamoto Y , Tsuchida H , Nakashima Y , Suzuki Y , Ikari H , Iguchi A ((1997) ) Pharmacokinetic study of aniracetam in elderly patients with cerebrovascular disease. Behav Brain Res 83: , 243–244.

[8] 

Kim CK , Lee YR , Ong L , Gold M , Kalali A , Sarkar J ((2022) ) Alzheimer’s disease: Key insights from two decades of clinical trial failures. J Alzheimers Dis 87: , 83–100.

[9] 

De Strooper B , Chávez Gutiérrez L ((2015) ) Learning by failing: Ideas and concepts to tackle γ-secretases in Alzheimer’s disease and beyond. Ann Rev Pharmacol Toxicol 55: , 419–437.

[10] 

Fahrenholz F ((2007) ) Alpha-secretase as a therapeutic target. Curr Alzheimer Res 4: , 412–417.

[11] 

Mattson MP , Cheng B , Culwell AR , Esch FS , Lieberburg I , Rydel RE ((1993) ) Evidence for excitoprotective and intraneuronal calcium-regulating roles for secreted forms of the β-amyloid precursor protein. Neuron 10: , 243–254.

[12] 

Furukawa K , Sopher BL , Rydel RE , Begley JG , Pham DG , Martin GM , Fox M Mattson MP ((1996) ) Increased activity-regulating and neuroprotective efficacy of α-secretase-derived secreted amyloid precursor protein conferred by a C-terminal heparin-binding domain. J Neurochem 67: , 1882–1896.

[13] 

Sastre M , Walter J , Gentleman SM ((2008) ) Interactions between APP secretases and inflammatory mediators. J Neuroinflammation 5: , 25.

[14] 

Akiyama H , Barger S , Barnum S , Bradt B , Bauer J , Cole GM , Cooper NR , Eikelenbloom P , Emmerling M , Fiebich BL , Finch CE , Frautschy S , Griffin WST , Hampel H , Hull M , Landreth G , Lue LF , Mrak R , Mackenzie IR , McGreer PL , O’Banion MK , Pachter J , Pasinetti G , Plata-Salaman C , Rogers J , Rydel R , Shen Y , Streit W , Stromeyer R , Tooyoma I , Van Muiswinkel FL , Veerhuis R , Walker D , Webster S , Wegrzyniak B , Wenk G , Wyss-Coray T ((2000) ) Inflammation and Alzheimer’s disease. Neurobiol Aging 21: , 383–421.

[15] 

Postina R ((2012) ) Activation of α-secretase cleavage. J Neurochem 120: , 46–54.

[16] 

Nigam SM , Xu S , Kritikou JS , Marosi K , Brodin L , Mattson MP ((2017) ) Exercise and BDNF reduce Aβ production by enhancing α-secretase processing of APP. J Neurochem 142: , 286–296.

[17] 

MacPherson RE ((2017) ) Filling the void: A role for exercise-induced BDNF and brain amyloid precursor protein processing. Am J Physiol Regul Integr Comp Physiol 313: , R585–R593.

[18] 

Gao L , Zhang Y , Sterling K , Song W ((2022) ) Brain-derived neurotrophic factor in Alzheimer’s disease and its pharmaceutical potential. Transl Neurodegener 11: , 4.

[19] 

Lee RK , Wurtman RJ , Cox AJ , Nitsch RM ((1995) ) Amyloid precursor protein processing is stimulated by metabotropic glutamate receptors. Proc Natl Acad Sci U S A 92: , 8083–8087.

[20] 

Nagele RG , Wegiel J , Venkataraman V , Imaki H , Wang KC , Wegiel J ((2004) ) Contribution of glial cells to the development of amyloid plaques in Alzheimer’s disease. Neurobiol Aging 25: , 663–674.

[21] 

Thinakaran G , Koo EH ((2008) ) Amyloid precursor protein trafficking, processing, and function. J Biol Chem 283: , 29615–29619.

[22] 

D’Andrea MR , Reiser PA , Gumula NA , Hertzog BM , Andrade-Gordon P ((2001) ) Application of triple immunohistochemistry to characterize amyloid plaque-associated inflammation in brains with Alzheimer’s disease. Biotech Histochem 76: , 97–106.

[23] 

LaFerla FM , Green KN , Oddo S ((2007) ) Intracellular amyloid-β in Alzheimer’s disease. Nat Rev Neurosci 8: , 499–509.

[24] 

Yankner BA ((1996) ) Mechanisms of neuronal degeneration in Alzheimer’s disease. Neuron 16: , 921–932.

[25] 

Tanila H ((2017) ) The role of BDNF in Alzheimer’s disease. Neurobiol Dis 97: , 114–118.

[26] 

Giovannini MG , Rodinò P , Mutolo D , Pepeu G ((1993) ) Oxiracetam and aniracetam increase acetylcholine release from the rat hippocampus in vivo. Drug Dev Res 28: , 503–509.

[27] 

Finder VH , Glockshuber R ((2007) ) Amyloid-β aggregation. Neurodegener Dis 4: , 13–27.

[28] 

Bredesen D ((2017) ) The end of Alzheimer’s: The first program to prevent and reverse cognitive decline. Penguin.

[29] 

Obregon D , Hou H , Deng J , Giunta B , Tian J , Darlington D , Shahaduzzaman M , Zhu Y , More T , Mattson MP , Tan J ((2012) ) Soluble amyloid precursor protein-α modulates β-secretase activity and amyloid-β generation. Nat Commun 3: , 777.

[30] 

Dar NJ , Glazner GW ((2020) ) Deciphering the neuroprotective and neurogenic potential of soluble amyloid precursor protein alpha (sAPPα). Cell Mol Life Sci 77: , 2315–2330.

[31] 

Tyler SJ , Dawbarn D , Wilcock GK , Allen SJ ((2002) ) α-and β-secretase: Profound changes in Alzheimer’s disease. Biochem Biophys Res Commun 299: , 373–376.

[32] 

Lichtenthaler SF , Haass C ((2004) ) Amyloid at the cutting edge: Activation of α-secretase prevents amyloidogenesis in an Alzheimer disease mouse model. J Clin Invest 113: , 1384–1387.

[33] 

Postina R ((2008) ) A closer look at α-secretase. Curr Alzheimer Res 5: , 179–186.

[34] 

Yiannopoulou KG , Papageorgiou SG ((2020) ) Current and future treatments in Alzheimer disease: An update. J Cent Nerv Syst Dis 12: , 1179573520907397.

[35] 

Ito I , Tanabe S , Kohda A , Sugiyama H ((1990) ) Allosteric potentiation of quisqualate receptors by a nootropic drug aniracetam. J Physiol 424: , 533–543.

[36] 

Tang CM , Shi QY , Katchman A , Lynch G ((1991) ) Modulation of the time course of fast EPSCs and glutamate channel kinetics by aniracetam. Science 254,: , 288–290.

[37] 

Copani A , Genazzani AA , Aleppo G , Casabona G , Canonico PL , Scapagnini U , Nicoletti F ((1992) ) Nootropic drugs positively modulate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-sensitive glutamate receptors in neuronal cultures. J Neurochem 58: , 1199–1204.

[38] 

Ahmed AH , Oswald RE ((2010) ) Piracetam defines a new binding site for allosteric modulators of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptors. J Med Chem 53,: , 2197–2203.

[39] 

Xie Y , Huang X , Hu SY , Zhang YJ , Wang Y , Qiu XJ , Ren P , Fan R , Zhang CH , Xie WB , Ji H , He J , Chen X , Xie L , Liu ZQ , Zhou HH ((2013) ) The involvement of AMPA–ERK1/2–BDNF pathway in the mechanism of new antidepressant action of prokinetic meranzin hydrate. Amino Acids 44: , 413–422.

[40] 

Xiao P , Staubli U , Kessler M , Lynch G ((1991) ) Selective effects of aniracetam across receptor types and forms of synaptic facilitation in hippocampus. Hippocampus 1: , 373–380.

[41] 

Vaglenova J , Pandiella N , Wijayawardhane N , Vaithianathan T , Birru S , Breese C , Suppiramaniam V , Randal C ((2008) ) Aniracetam reversed learning and memory deficits following prenatal ethanol exposure by modulating functions of synaptic AMPA receptors. Neuropsychopharmacology 33: , 1071–1083.

[42] 

Al Khashali H , Ray R , Coleman KL , Atali S , Haddad B , Wareham J , Guthrie J , Heyl D , Evans HG ((2022) ) Regulation of the soluble amyloid precursor protein α (sAPPα) levels by acetylcholinesterase and brain-derived neurotrophic factor in lung cancer cell media. Int J Mol Sci 23: , 10746.

[43] 

Holback S , Adlerz L , Iverfeldt K ((2005) ) Increased processing of APLP2 and APP with concomitant formation of APP intracellular domains in BDNF and retinoic acid-differentiated human neuroblastoma cells. J Neurochem 95: , 1059–1068.

[44] 

Laske C , Stransky E , Leyhe T , Eschweiler GW , Maetzler W , Wittorf A , Soekadar S , Richartz E , Koehler N , Bartels M , Buchkremer G , Schott K ((2007) ) BDNF serum and CSF concentrations in Alzheimer’s disease, normal pressure hydrocephalus and healthy controls. J Psychiatr Res 41: , 387–394.

[45] 

Takeda K , Kermani P , Anastasia A , Obinata Y , Hempstead BL , Kurihara H ((2013) ) BDNF protects human vascular endothelial cells from TNFα-induced apoptosis. Biochem Cell Biol 91: , 341–349.

[46] 

Wu CL , Hwang CS , Chen SD , Yin JH , Yang DI ((2010) ) Neuroprotective mechanisms of brain-derived neurotrophic factor against 3-nitropropionic acid toxicity: Therapeutic implications for Huntington’s disease. Ann N Y Acad Sci 1201: , 8–12.

[47] 

Swain M , K Soman S , Tapia K , Dagda RY , Dagda RK ((2023) ) Brain-derived neurotrophic factor protects neurons by stimulating mitochondrial function through protein kinase A. J Neurochem 167: , 104–125.

[48] 

Hirai K , Aliev G , Nunomura A , Fujioka H , Russell RL , Atwood CS , Johnson AB , Kress Y , Vinters HV , Tabaton M , Shimohama S , Cash AD , Siedlak SL , Harris PLR , Jones PK , Petersen RB , Perry G , Smith MA ((2001) ) Mitochondrial abnormalities in Alzheimer’s disease. J Neurosci 21: , 3017–3023.

[49] 

Bell SM , Barnes K , De Marco M , Shaw PJ , Ferraiuolo L , Blackburn DJ , Venneri A Mortiboys H ((2021) ) Mitochondrial dysfunction in Alzheimer’s disease: A biomarker of the future? Biomedicines 9: , 63.

[50] 

Huang WJ , Zhang XIA. , Chen WW ((2016) ) Role of oxidative stress in Alzheimer’s disease. Biomed Rep 4: , 519–522.

[51] 

Peng S , Wuu J , Mufson EJ , Fahnestock M ((2005) ) Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer’s disease. J Neurochem 93: , 1412–1421.

[52] 

Lee HG , Zhu X , O’Neill M , Webber K , Casadesus G , Marlatt M , Raina A , Perry G , Smith M ((2004) ) The role of metabotropic glutamate receptors in Alzheimer’s disease. Acta Neurobiol Exp 64: , 89–98.

[53] 

Srivastava A , Das B , Yao AY , Yan R ((2020) ) Metabotropic glutamate receptors in Alzheimer’s disease synaptic dysfunction: Therapeutic opportunities and hope for the future. J Alzheimers Dis 78: , 1345–1361.

[54] 

Deng W , Wang H , Rosenberg PA , Volpe JJ , Jensen FE ((2004) ) Role of metabotropic glutamate receptors in oligodendrocyte excitotoxicity and oxidative stress. Proc Natl Acad Sci U S A 101: , 7751–7756.

[55] 

Pizzi M , Consolandi O , Memo M , Spano P ((1996) ) Activation of multiple metabotropic glutamate receptor subtypes prevents NMDA-induced excitotoxicity in rat hippocampal slices. Eur J Neurosci 8: , 1516–1521.

[56] 

Kim SH , Fraser PE , Westaway D , George-Hyslop PHS , Ehrlich ME , Gandy S ((2010) ) Group II metabotropic glutamate receptor stimulation triggers production and release of Alzheimer’s amyloid β42 from isolated intact nerve terminals. J Neurosci 30: , 3870–3875.

[57] 

Thathiah A , De Strooper B ((2011) ) The role of G protein-coupled receptors in the pathology of Alzheimer’s disease. Nat Rev Neurosci 12: , 73–87.

[58] 

Durand D , Carniglia L , Turati J , Ramírez D , Saba J , Caruso C , Lasaga M ((2017) ) Amyloid-beta neurotoxicity and clearance are both regulated by glial group II metabotropic glutamate receptors. Neuropharmacology 123: , 274–286.

[59] 

Durand D , Carniglia L , Beauquis J , Caruso C , Saravia F , Lasaga M ((2014) ) Astroglial mGlu3 receptors promote alpha-secretase-mediated amyloid precursor protein cleavage. Neuropharmacology 79: , 180–189.

[60] 

Kumar A , Dhull DK , Mishra PS ((2015) ) Therapeutic potential of mGluR5 targeting in Alzheimer’s disease. Front Neurosci 9: , 215.

[61] 

Senin U , Abate G , Fieschi C , Gori G , Guala A , Marini G , Villardita C Parnetti L ((1991) ) Aniracetam (Ro 13-5057) in the treatment of senile dementia of Alzheimer type (SDAT): Results of a placebo controlled multicentre clinical study. Eur Neuropsychopharmacol 1: , 511–517.

[62] 

Parnetti L , Bartorelli L , Bonaiuto S , Cucinotta D , Cuzzupoli M , Erminio F , Maggioni M , Marini G , Paciaroni E , Pedrazzi F , Peruzza M , Senin U ((1991) ) Aniracetam (Ro 13-5057) for the treatment of senile dementia of Alzheimer type: Results of a multicentre clinical study. Dement Geriatr Cogn Disord 2: , 262–267.

[63] 

Wei W , Norton DD , Wang X , Kusiak JW ((2002) ) Aβ 17–42 in Alzheimer’s disease activates JNK and caspase-8 leading to neuronal apoptosis. Brain 125: , 2036–2043.

[64] 

Kuhn AJ , Abrams BS , Knowlton S , Raskatov JA ((2020) ) Alzheimer’s disease “non-amyloidogenic” p3 peptide revisited: A case for amyloid-α . ACS Chem Neurosci 11: , 1539–1544.

[65] 

Canonico V , Forgione L , Paoletti C , Casini A , Colonna CV , Bertini M , Acito R , Rengo F ((1991) ) Efficacy and tolerance of aniracetam in elderly patients with primary or secondary mental deterioration. Riv Neurol 61: , 92–96.

[66] 

Koliaki CC , Messini C , Tsolaki M ((2012) ) Clinical efficacy of aniracetam, either as monotherapy or combined with cholinesterase inhibitors, in patients with cognitive impairment: A comparative open study. CNS Neurosci Therap 18: , 302–312.